Skip to main content

Patient-Derived In Vitro and In Vivo Models of Cancer

  • Chapter
  • First Online:
Computational Methods for Precision Oncology

Abstract

Over the last two decades, cancer researchers have taken the promise offered by the Human Genome Project and have expanded its capacity to use sequencing to identify the genomic alterations that give rise to and sustain individual tumors. This expansion has allowed researchers to identify and target highly recurrent alterations in specific cancer contexts, such as EGFR mutations in non-small cell lung cancer (Lynch et al, N Engl J Med 350:2129–2139, 2004; Sharifnia et al., Proc Natl Acad Sci U S A 111:18661–18666, 2014), BCR-ABL translocations in chronic myeloid leukemia (Deininger, Pharmacol Rev 55:401–423. https://doi.org/10.1124/pr.55.3.4, 2003; Druker et al, N Engl J Med 344. 1038–1042, 2001; Druker et al, N Engl J Med 344:1031–1037. https://doi.org/10.1056/NEJM200104053441401, 2001), or HER2 amplifications in breast cancer (Slamon et al, N Engl J Med 344:783–792. https://doi.org/10.1056/NEJM200103153441101, 2001; Solca et al, Beyond trastuzumab: second-generation targeted therapies for HER-2-positive breast cancer. In: Sibilia M, Zielinski CC, Bartsch R, Grunt TW (eds) Drugs for HER-2-positive breast cancer. Springer, Basel, pp 91–107, 2011). Despite these advances in our capacity to identify the genetic alterations that drive tumor initiation, survival, and proliferation, our ability to target these alterations to provide effective treatment options for patients in need, particularly those with rare or advanced cancers, remains limited (Gould et al, Nat Med 21:431–439. https://doi.org/10.1038/nm.3853, 2015). Patient-derived models of cancer offer one potential mechanism to overcome this barrier between the bench and bedside. Through the development and testing of patient-derived models of cancer, functional genomics efforts can identify tumor-specific drug sensitivities and thereby provide a connection between tumor genetics and effective therapeutics for patients in need of treatment options.

Recognizing that cancer is a multifaceted set of disease states, the development of personalized models of cancer that can be used to compare treatment options, identify tumor-specific vulnerabilities, and guide clinical decision-making has tremendous potential for improving patient outcomes. This chapter will describe a representative set of patient-derived models of cancer, reviewing each of their strengths and weaknesses and highlighting how selecting a model to suit a specific question or context is critical. Each model comes with a unique set of pros and cons, making them more or less appropriate for each specific research or clinical question. As each model can be leveraged to gain new insights into cancer biology, the key to their deployment is to identify the most appropriate model for a specific context, while carefully considering the strengths and limitations of the selected model. When used appropriately, patient-derived models may prove to be the missing link needed to bring the promise of personalized oncology to fruition in the clinic.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 169.00
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Hardcover Book
USD 219.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

References

  1. Gould SE, Junttila MR, de Sauvage FJ. Translational value of mouse models in oncology drug development. Nat Med. 2015;21:431–9. https://doi.org/10.1038/nm.3853.

    Article  CAS  PubMed  Google Scholar 

  2. Klemm F, Joyce JA. Microenvironmental regulation of therapeutic response in cancer. Trends Cell Biol. 2015;25:198–213. https://doi.org/10.1016/j.tcb.2014.11.006.

    Article  PubMed  Google Scholar 

  3. Olive KP, Jacobetz MA, Davidson CJ, Gopinathan A, McIntyre D, Honess D, Madhu B, Goldgraben MA, Caldwell ME, Allard D, Frese KK, DeNicola G, Feig C, Combs C, Winter SP, Ireland-Zecchini H, Reichelt S, Howat WJ, Chang A, Dhara M, Wang L, Rückert F, Grützmann R, Pilarsky C, Izeradjene K, Hingorani SR, Huang P, Davies SE, Plunkett W, Egorin M, Hruban RH, Whitebread N, McGovern K, Adams J, Iacobuzio-Donahue C, Griffiths J, Tuveson DA. Inhibition of hedgehog signaling enhances delivery of chemotherapy in a mouse model of pancreatic cancer. Science. 2009;324:1457–61. https://doi.org/10.1126/science.1171362.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Podsypanina K, Ellenson LH, Nemes A, Gu J, Tamura M, Yamada KM, Cordon-Cardo C, Catoretti G, Fisher PE, Parsons R. Mutation of Pten/Mmac1 in mice causes neoplasia in multiple organ systems. Proc Natl Acad Sci. 1999;96:1563–8. https://doi.org/10.1073/pnas.96.4.1563.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Sos ML, Michel K, Zander T, Weiss J, Frommolt P, Peifer M, Li D, Ullrich R, Koker M, Fischer F, Shimamura T, Rauh D, Mermel C, Fischer S, Stückrath I, Heynck S, Beroukhim R, Lin W, Winckler W, Shah K, LaFramboise T, Moriarty WF, Hanna M, Tolosi L, Rahnenführer J, Verhaak R, Chiang D, Getz G, Hellmich M, Wolf J, Girard L, Peyton M, Weir BA, Chen T-H, Greulich H, Barretina J, Shapiro GI, Garraway LA, Gazdar AF, Minna JD, Meyerson M, Wong K-K, Thomas RK. Predicting drug susceptibility of non–small cell lung cancers based on genetic lesions. J Clin Invest. 2009;119:1727–40. https://doi.org/10.1172/JCI37127.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Lynch TJ, Well DW, Sordella R, Gurubhagavatula S, Okimoto RA, Brannigan BW, Harris PL, Haserlat SM, Supko JG, Haluska FG, Louis DN, Christiani DC, Settleman J, Haber DA. Activating mutations in the epidermal growth factor receptor underlying responsiveness of non–small-cell lung cancer to gefitinib. N Engl J Med. 2004;350:2129–39. https://doi.org/10.1056/NEJMoa040938.

    Article  CAS  PubMed  Google Scholar 

  7. Sharifnia T, Rusu V, Piccioni F, Bagul M, Imielinski M, Cherniack AD, Pedamallu CS, Wong B, Wilson FH, Garraway LA, Altshuler D, Golub TR, Root DE, Subramanian A, Meyerson M. Genetic modifiers of EGFR dependence in non-small cell lung cancer. Proc Natl Acad Sci U S A. 2014;111:18661–6. https://doi.org/10.1073/pnas.1412228112.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Deininger MWN. Specific targeted therapy of chronic myelogenous leukemia with imatinib. Pharmacol Rev. 2003;55:401–23. https://doi.org/10.1124/pr.55.3.4.

    Article  CAS  PubMed  Google Scholar 

  9. Druker BJ, Sawyers CL, Kantarjian H, Resta DJ, Reese SF, Ford JM, Capdeville R, Talpaz M. Activity of a specific inhibitor of the BCR-ABL tyrosine kinase in the blast crisis of chronic myeloid leukemia and acute lymphoblastic leukemia with the Philadelphia chromosome. N Engl J Med. 2001;344:1038–42. https://doi.org/10.1056/NEJM200104053441402.

    Article  CAS  PubMed  Google Scholar 

  10. Druker BJ, Talpaz M, Resta DJ, Peng B, Buchdunger E, Ford JM, Lydon NB, Kantarjian H, Capdeville R, Ohno-Jones S, Sawyers CL. Efficacy and safety of a specific inhibitor of the BCR-ABL tyrosine kinase in chronic myeloid leukemia. N Engl J Med. 2001;344:1031–7. https://doi.org/10.1056/NEJM200104053441401.

    Article  CAS  PubMed  Google Scholar 

  11. Slamon DJ, Leyland-Jones B, Shak S, Fuchs H, Paton V, Bajamonde A, Fleming T, Eiermann W, Wolter J, Pegram M, Baselga J, Norton L. Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N Engl J Med. 2001;344:783–92. https://doi.org/10.1056/NEJM200103153441101.

    Article  CAS  PubMed  Google Scholar 

  12. Solca FF, Adolf GR, Jones H, Uttenreuther-Fischer MM. Beyond trastuzumab: second-generation targeted therapies for HER-2-positive breast cancer. In: Sibilia M, Zielinski CC, Bartsch R, Grunt TW, editors. Drugs for HER-2-positive breast cancer. Basel: Springer; 2011. p. 91–107. https://doi.org/10.1007/978-3-0346-0094-1_6.

  13. Shaw AT, Friboulet L, Leshchiner I, Gainor JF, Bergqvist S, Brooun A, Burke BJ, Deng Y-L, Liu W, Dardaei L, Frias RL, Schultz KR, Logan J, James LP, Smeal T, Timofeevski S, Katayama R, Iafrate AJ, Le L, McTigue M, Getz G, Johnson TW, Engelman JA. Resensitization to crizotinib by the lorlatinib ALK resistance mutation L1198F. N Engl J Med. 2016;374:54–61. https://doi.org/10.1056/NEJMoa1508887.

    Article  CAS  PubMed  Google Scholar 

  14. Johannessen CM, Boehm JS. Progress towards precision functional genomics in cancer. Curr Opin Syst Biol. 2017;2:74–83. https://doi.org/10.1016/j.coisb.2017.02.002.

    Article  Google Scholar 

  15. Letai A. Functional precision cancer medicine—moving beyond pure genomics. Nat Med. 2017;23:1028–35. https://doi.org/10.1038/nm.4389.

    Article  CAS  PubMed  Google Scholar 

  16. Tyner JW. Integrating functional genomics to accelerate mechanistic personalized medicine. Cold Spring Harb Mol Case Stud. 2017;3:a001370. https://doi.org/10.1101/mcs.a001370.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Alley MC, Scudiere DA, Monks A, Hursey ML, Czerwinski MJ, Fine DL, Abbott BJ, Mayo JG, Shoemaker RH, Boyd MR. Feasibility of drug screening with panels of human tumor cell lines using a microculture tetrazolium assay. Cancer Res. 1988;48:589–601. PMID: 3335022.

    CAS  PubMed  Google Scholar 

  18. Stinson S, Alley M, Kopp W, Fiebig H, Mullendore L, Pittman A, Kenney S, Keller J, Boyd M. Morphological and immunocytochemical characteristics of human tumor cell lines for use in a disease-oriented anticancer drug screen. Anticancer Res. 1992;12:1035–53. PMID: 1503399.

    CAS  PubMed  Google Scholar 

  19. Bairoch A. The cellosaurus, a cell-line knowledge resource. J Biomol Tech. 2018;29:25–38. https://doi.org/10.7171/jbt.18-2902-002.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Greshock J, Bachman KE, Degenhardt YY, Jing J, Wen YH, Eastman S, McNeil E, Moy C, Wegrzyn R, Auger K, Hardwicke MA, Wooster R. Molecular target class is predictive of in vitro response profile. Cancer Res. 2010;70:3677–86. https://doi.org/10.1158/0008-5472.CAN-09-3788.

    Article  CAS  PubMed  Google Scholar 

  21. Heiser LM, Sadanandam A, Kuo W-L, Benz SC, Goldstein TC, Ng S, Gibb WJ, Wang NJ, Ziyad S, Tong F, Bayani N, Hu Z, Billig JI, Dueregger A, Lewis S, Jakkula L, Korkola JE, Durinck S, Pepin F, Guan Y, Purdom E, Neuvial P, Bengtsson H, Wood KW, Smith PG, Vassilev LT, Hennessy BT, Greshock J, Bachman KE, Hardwicke MA, Park JW, Marton LJ, Wolf DM, Collisson EA, Neve RM, Mills GB, Speed TP, Feiler HS, Wooster RF, Haussler D, Stuart JM, Gray JW, Spellman PT. Subtype and pathway specific responses to anticancer compounds in breast cancer. Proc Natl Acad Sci U S A. 2012;109:2724–9. https://doi.org/10.1073/pnas.1018854108.

  22. Marcotte R, Sayad A, Brown KR, Sanchez-Garcia F, Reimand J, Haider M, Virtanen C, Bradner JE, Bader GD, Mills GB, Pe’er D, Moffat J, Neel BG. Functional genomic landscape of human breast cancer drivers, vulnerabilities, and resistance. Cell. 2016;164:293–309. https://doi.org/10.1016/j.cell.2015.11.062.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. McDonald ER, de Weck A, Schlabach MR, Billy E, Mavrakis KJ, Hoffman GR, Belur D, Castelletti D, Frias E, Gampa K, Golji J, Kao I, Li L, Megel P, Perkins TA, Ramadan N, Ruddy DA, Silver SJ, Sovath S, Stump M, Weber O, Widmer R, Yu J, Yu K, Yue Y, Abramowski D, Ackley E, Barrett R, Berger J, Bernard JL, Billig R, Brachmann SM, Buxton F, Caothien R, Caushi JX, Chung FS, Cortés-Cros M, deBeaumont RS, Delaunay C, Desplat A, Duong W, Dwoske DA, Eldridge RS, Farsidjani A, Feng F, Feng J, Flemming D, Forrester W, Galli GG, Gao Z, Gauter F, Gibaja V, Haas K, Hattenberger M, Hood T, Hurov KE, Jagani Z, Jenal M, Johnson JA, Jones MD, Kapoor A, Korn J, Liu J, Liu Q, Liu S, Liu Y, Loo AT, Macchi KJ, Martin T, McAllister G, Meyer A, Mollé S, Pagliarini RA, Phadke T, Repko B, Schouwey T, Shanahan F, Shen Q, Stamm C, Stephan C, Stucke VM, Tiedt R, Varadarajan M, Venkatesan K, Vitari AC, Wallroth M, Weiler J, Zhang J, Mickanin C, Myer VE, Porter JA, Lai A, Bitter H, Lees E, Keen N, Kauffmann A, Stegmeier F, Hofmann F, Schmelzle T, Sellers WR. Project DRIVE: a compendium of cancer dependencies and synthetic lethal relationships uncovered by large-scale, deep RNAi screening. Cell. 2017;170:577–592.e10. https://doi.org/10.1016/j.cell.2017.07.005.

    Article  CAS  PubMed  Google Scholar 

  24. McFarland JM, Ho ZV, Kugener G, Dempster JM, Montgomery PG, Bryan JG, Krill-Burger JM, Green TM, Vazquez F, Boehm JS, Golub TR, Hahn WC, Root DE, Tsherniak A. Improved estimation of cancer dependencies from large-scale RNAi screens using model-based normalization and data integration. Nat Commun. 2018;9:1–13. https://doi.org/10.1038/s41467-018-06916-5.

    Article  CAS  Google Scholar 

  25. Meyers RM, Bryan JG, McFarland JM, Weir BA, Sizemore AE, Xu H, Dharia NV, Montgomery PG, Cowley GS, Pantel S, Goodale A, Lee Y, Ali LD, Jiang G, Lubonja R, Harrington WF, Strickland M, Wu T, Hawes DC, Zhivich VA, Wyatt MR, Kalani Z, Chang JJ, Okamoto M, Stegmaier K, Golub TR, Boehm JS, Vazquez F, Root DE, Hahn WC, Tsherniak A. Computational correction of copy-number effect improves specificity of CRISPR-Cas9 essentiality screens in cancer cells. Nat Genet. 2017;49:1779–84. https://doi.org/10.1038/ng.3984.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Tsherniak A, Vazquez F, Montgomery PG, Weir BA, Kryukov G, Cowley GS, Gill S, Harrington WF, Pantel S, Krill-Burger JM, Meyers RM, Ali L, Goodale A, Lee Y, Jiang G, Hsiao J, Gerath WFJ, Howell S, Merkel E, Ghandi M, Garraway LA, Root DE, Golub TR, Boehm JS, Hahn WC. Defining a cancer dependency map. Cell. 2017;170:564–576.e16. https://doi.org/10.1016/j.cell.2017.06.010.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Yu C, Mannan AM, Yvone GM, Ross KN, Zhang Y-L, Marton MA, Taylor BR, Crenshaw A, Gould JZ, Tamayo P, Weir BA, Tsherniak A, Wong B, Garraway LA, Shamji AF, Palmer MA, Foley MA, Winckler W, Schreiber SL, Kung AL, Golub TR. High-throughput identification of genotype-specific cancer vulnerabilities in mixtures of barcoded tumor cell lines. Nat Biotechnol. 2016;34:419–23. https://doi.org/10.1038/nbt.3460.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Barretina J, Caponigro G, Stransky N, Venkatesan K, Margolin AA, Kim S, Wilson CJ, Lehár J, Kryukov GV, Sonkin D, Reddy A, Liu M, Murray L, Berger MF, Monahan JE, Morais P, Meltzer J, Korejwa A, Jané-Valbuena J, Mapa FA, Thibault J, Bric-Furlong E, Raman P, Shipway A, Engels IH, Cheng J, Yu GK, Yu J, Aspesi P, de Silva M, Jagtap K, Jones MD, Wang L, Hatton C, Palescandolo E, Gupta S, Mahan S, Sougnez C, Onofrio RC, Liefeld T, MacConaill L, Winckler W, Reich M, Li N, Mesirov JP, Gabriel SB, Getz G, Ardlie K, Chan V, Myer VE, Weber BL, Porter J, Warmuth M, Finan P, Harris JL, Meyerson M, Golub TR, Morrissey MP, Sellers WR, Schlegel R, Garraway LA. The Cancer Cell Line Encyclopedia enables predictive modelling of anticancer drug sensitivity. Nature. 2012;483:603–7. https://doi.org/10.1038/nature11003.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Cancer Cell Line Encyclopedia Consortium, Genomics of Drug Sensitivity in Cancer Consortium. Pharmacogenomic agreement between two cancer cell line data sets. Nature. 2015;528:84–7. https://doi.org/10.1038/nature15736.

    Article  CAS  Google Scholar 

  30. Garnett MJ, Edelman EJ, Heidorn SJ, Greenman CD, Dastur A, Lau KW, Greninger P, Thompson IR, Luo X, Soares J, Liu Q, Iorio F, Surdez D, Chen L, Milano RJ, Bignell GR, Tam AT, Davies H, Stevenson JA, Barthorpe S, Lutz SR, Kogera F, Lawrence K, McLaren-Douglas A, Mitropoulos X, Mironenko T, Thi H, Richardson L, Zhou W, Jewitt F, Zhang T, O’Brien P, Boisvert JL, Price S, Hur W, Yang W, Deng X, Butler A, Choi HG, Chang JW, Baselga J, Stamenkovic I, Engelman JA, Sharma SV, Delattre O, Saez-Rodriguez J, Gray NS, Settleman J, Futreal PA, Haber DA, Stratton MR, Ramaswamy S, McDermott U, Benes CH. Systematic identification of genomic markers of drug sensitivity in cancer cells. Nature. 2012;483:570–5. https://doi.org/10.1038/nature11005.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Yang W, Soares J, Greninger P, Edelman EJ, Lightfoot H, Forbes S, Bindal N, Beare D, Smith JA, Thompson IR, Ramaswamy S, Futreal PA, Haber DA, Stratton MR, Benes C, McDermott U, Garnett MJ. Genomics of Drug Sensitivity in Cancer (GDSC): a resource for therapeutic biomarker discovery in cancer cells. Nucleic Acids Res. 2012;41:D955–61. https://doi.org/10.1093/nar/gks1111.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Basu A, Bodycombe NE, Cheah JH, Price EV, Liu K, Schaefer GI, Ebright RY, Stewart ML, Ito D, Wang S, Bracha AL, Liefeld T, Wawer M, Gilbert JC, Wilson AJ, Stransky N, Kryukov GV, Dancik V, Barretina J, Garraway LA, Hon CS, Munoz B, Bittker JA, Stockwell BR, Khabele D, Stern AM, Clemons PA, Shamji AF, Schreiber SL. An interactive resource to identify cancer genetic and lineage dependencies targeted by small molecules. Cell. 2013;154:1151–61. https://doi.org/10.1016/j.cell.2013.08.003.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Rees MG, Seashore-Ludlow B, Cheah JH, Adams DJ, Price EV, Gill S, Javaid S, Coletti ME, Jones VL, Bodycombe NE, Soule CK, Alexander B, Li A, Montgomery P, Kotz JD, Hon CS-Y, Munoz B, Liefeld T, Dančík V, Haber DA, Clish CB, Bittker JA, Palmer M, Wagner BK, Clemons PA, Shamji AF, Schreiber SL. Correlating chemical sensitivity and basal gene expression reveals mechanism of action. Nat Chem Biol. 2016;12:109–16. https://doi.org/10.1038/nchembio.1986.

    Article  CAS  PubMed  Google Scholar 

  34. Seashore-Ludlow B, Rees MG, Cheah JH, Cokol M, Price EV, Coletti ME, Jones V, Bodycombe NE, Soule CK, Gould J, Alexander B, Li A, Montgomery P, Wawer MJ, Kuru N, Kotz JD, Hon CS, Munoz B, Liefeld T, Dančík V, Bittker JA, Palmer M, Bradner JE, Shamji AF, Clemons PA, Schreiber SL. Harnessing connectivity in a large-scale small-molecule sensitivity dataset. Cancer Discov. 2015;5:1210–23. https://doi.org/10.1158/2159-8290.CD-15-0235.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Ben-David U, Siranosian B, Ha G, Tang H, Oren Y, Hinohara K, Strathdee CA, Dempster J, Lyons NJ, Burns R, Nag A, Kugener G, Cimini B, Tsvetkov P, Maruvka YE, O’Rourke R, Garrity A, Tubelli AA, Bandopadhayay P, Tsherniak A, Vazquez F, Wong B, Birger C, Ghandi M, Thorner AR, Bittker JA, Meyerson M, Getz G, Beroukhim R, Golub TR. Genetic and transcriptional evolution alters cancer cell line drug response. Nature. 2018;560:325–30. https://doi.org/10.1038/s41586-018-0409-3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Haibe-Kains B, El-Hachem N, Birkbak NJ, Jin AC, Beck AH, Aerts HJWL, Quackenbush J. Inconsistency in large pharmacogenomic studies. Nature. 2013;504:389–93. https://doi.org/10.1038/nature12831.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Jang IS, Neto EC, Guinney J, Friend SH, Margolin AA. Systematic assessment of analytical methods for drug sensitivity prediction from cancer cell line data. Pac Symp Biocomput. 2014:63–74. PMID: 24297534; PMCID: PMC3995541.

    Google Scholar 

  38. Geeleher P, Cox NJ, Huang RS. Cancer biomarker discovery is improved by accounting for variability in general levels of drug sensitivity in pre-clinical models. Genome Biol. 2016;17:190. https://doi.org/10.1186/s13059-016-1050-9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Geeleher P, Gamazon ER, Seoighe C, Cox NJ, Huang RS. Consistency in large pharmacogenomic studies. Nature. 2016;540:E1–2. https://doi.org/10.1038/nature19838.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Hatzis C, Bedard PL, Juul Birkbak N, Beck AH, Aerts HJWL, Stern DF, Shi L, Clarke R, Quackenbush J, Haibe-Kains B. Enhancing reproducibility in cancer drug screening: how do we move forward? Cancer Res. 2014;74:4016–23. https://doi.org/10.1158/0008-5472.CAN-14-0725.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Haverty PM, Lin E, Tan J, Yu Y, Lam B, Lianoglou S, Neve RM, Martin S, Settleman J, Yauch RL, Bourgon R. Reproducible pharmacogenomic profiling of cancer cell line panels. Nature. 2016;533:333–7. https://doi.org/10.1038/nature17987.

    Article  CAS  PubMed  Google Scholar 

  42. Safikhani Z, El-Hachem N, Smirnov P, Freeman M, Goldenberg A, Birkbak NJ, Beck AH, Aerts HJWL, Quackenbush J, Haibe-Kains B. Safikhani et al. reply. Nature. 2016;540:E2–4. https://doi.org/10.1038/nature19839.

    Article  CAS  PubMed  Google Scholar 

  43. Safikhani Z, Smirnov P, Freeman M, El-Hachem N, She A, Rene Q, Goldenberg A, Birkbak NJ, Hatzis C, Shi L, Beck AH, Aerts HJWL, Quackenbush J, Haibe-Kains B. Revisiting inconsistency in large pharmacogenomic studies. F1000Research. 2017;5:2333. https://doi.org/10.12688/f1000research.9611.3.

    Article  PubMed Central  Google Scholar 

  44. Pauli C, Hopkins BD, Prandi D, Shaw R, Fedrizzi T, Sboner A, Sailer V, Augello M, Puca L, Rosati R, McNary TJ, Churakova Y, Cheung C, Triscott J, Pisapia D, Rao R, Mosquera JM, Robinson B, Faltas BM, Emerling BE, Gadi VK, Bernard B, Elemento O, Beltran H, Demichelis F, Kemp CJ, Grandori C, Cantley LC, Rubin MA. Personalized in vitro and in vivo cancer models to guide precision medicine. Cancer Discov. 2017;7:462–77. https://doi.org/10.1158/2159-8290.CD-16-1154.

    Article  PubMed  PubMed Central  Google Scholar 

  45. Evrard YA, Srivastava A, Randjelovic J, Doroshow JH, Dean DA, Morris JS, Chuang JH. Systematic establishment of robustness and standards in patient-derived xenograft experiments and analysis. Cancer Res. 2020;80:2286–97. https://doi.org/10.1158/0008-5472.CAN-19-3101.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Meehan TF, Conte N, Goldstein T, Inghirami G, Murakami MA, Brabetz S, Gu Z, Wiser JA, Dunn P, Begley DA, Krupke DM, Bertotti A, Bruna A, Brush MH, Byrne AT, Caldas C, Christie AL, Clark DA, Dowst H, Dry JR, Doroshow JH, Duchamp O, Evrard YA, Ferretti S, Frese KK, Goodwin NC, Greenawalt D, Haendel MA, Hermans E, Houghton PJ, Jonkers J, Kemper K, Khor TO, Lewis MT, Lloyd KCK, Mason J, Medico E, Neuhauser SB, Olson JM, Peeper DS, Rueda OM, Seong JK, Trusolino L, Vinolo E, Wechsler-Reya RJ, Weinstock DM, Welm A, Weroha SJ, Amant F, Pfister SM, Kool M, Parkinson H, Butte AJ, Bult CJ. PDX-MI: minimal information for patient-derived tumor xenograft models. Cancer Res. 2017;77:e62–6. https://doi.org/10.1158/0008-5472.CAN-17-0582.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Tatum JL, Kalen JD, Jacobs PM, Ileva LV, Riffle LA, Hollingshead MG, Doroshow JH. A spontaneously metastatic model of bladder cancer: imaging characterization. J Transl Med. 2019;17:425. https://doi.org/10.1186/s12967-019-02177-y.

    Article  PubMed  PubMed Central  Google Scholar 

  48. Yu M, Selvaraj SK, Liang-Chu MMY, Aghajani S, Busse M, Yuan J, Lee G, Peale F, Klijn C, Bourgon R, Kaminker JS, Neve RM. A resource for cell line authentication, annotation and quality control. Nature. 2015;520:307–11. https://doi.org/10.1038/nature14397.

    Article  CAS  PubMed  Google Scholar 

  49. Lee SH, Hu W, Matulay JT, Silva MV, Owczarek TB, Kim K, Chua CW, Barlow LJ, Kandoth C, Williams AB, Bergren SK, Pietzak EJ, Anderson CB, Benson MC, Coleman JA, Taylor BS, Abate-Shen C, McKiernan JM, Al-Ahmadie H, Solit DB, Shen MM. Tumor evolution and drug response in patient-derived organoid models of bladder cancer. Cell. 2018;173:515–528.e17. https://doi.org/10.1016/j.cell.2018.03.017.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Mullenders J, de Jongh E, Brousali A, Roosen M, Blom JPA, Begthel H, Korving J, Jonges T, Kranenburg O, Meijer R, Clevers HC. Mouse and human urothelial cancer organoids: a tool for bladder cancer research. Proc Natl Acad Sci. 2019;116:4567–74. https://doi.org/10.1073/pnas.1803595116.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Ballabio C, Anderle M, Gianesello M, Lago C, Miele E, Cardano M, Aiello G, Piazza S, Caron D, Gianno F, Ciolfi A, Pedace L, Mastronuzzi A, Tartaglia M, Locatelli F, Ferretti E, Giangaspero F, Tiberi L. Modeling medulloblastoma in vivo and with human cerebellar organoids. Nat Commun. 2020;11:583. https://doi.org/10.1038/s41467-019-13989-3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Linkous A, Balamatsias D, Snuderl M, Edwards L, Miyaguchi K, Milner T, Reich B, Cohen-Gould L, Storaska A, Nakayama Y, Schenkein E, Singhania R, Cirigliano S, Magdeldin T, Lin Y, Nanjangud G, Chadalavada K, Pisapia D, Liston C, Fine HA. Modeling patient-derived glioblastoma with cerebral organoids. Cell Rep. 2019;26:3203–3211.e5. https://doi.org/10.1016/j.celrep.2019.02.063.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Sachs N, de Ligt J, Kopper O, Gogola E, Bounova G, Weeber F, Balgobind AV, Wind K, Gracanin A, Begthel H, Korving J, van Boxtel R, Duarte AA, Lelieveld D, van Hoeck A, Ernst RF, Blokzijl F, Nijman IJ, Hoogstraat M, van de Ven M, Egan DA, Zinzalla V, Moll J, Boj SF, Voest EE, Wessels L, van Diest PJ, Rottenberg S, Vries RGJ, Cuppen E, Clevers H. A living biobank of breast cancer organoids captures disease heterogeneity. Cell. 2018;172:373–386.e10. https://doi.org/10.1016/j.cell.2017.11.010.

    Article  CAS  PubMed  Google Scholar 

  54. Mazzucchelli S, Piccotti F, Allevi R, Truffi M, Sorrentino L, Russo L, Agozzino M, Signati L, Bonizzi A, Villani L, Corsi F. Establishment and morphological characterization of patient-derived organoids from breast cancer. Biol Proced Online. 2019;21:1–3. https://doi.org/10.1186/s12575-019-0099-8.

    Article  Google Scholar 

  55. Goldhammer N, Kim J, Timmermans-Wielenga V, Petersen OW. Characterization of organoid cultured human breast cancer. Breast Cancer Res. 2019;21:141. https://doi.org/10.1186/s13058-019-1233-x.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Fujii M, Shimokawa M, Date S, Takano A, Matano M, Nanki K, Ohta Y, Toshimitsu K, Nakazato Y, Kawasaki K, Uraoka T, Watanabe T, Kanai T, Sato T. A colorectal tumor organoid library demonstrates progressive loss of niche factor requirements during tumorigenesis. Cell Stem Cell. 2016;18:827–38. https://doi.org/10.1016/j.stem.2016.04.003.

    Article  CAS  PubMed  Google Scholar 

  57. Narasimhan V, Wright JA, Churchill M, Wang T, Rosati R, Lannagan TRM, Vrbanac L, Richardson AB, Kobayashi H, Price T, Tye GXY, Marker J, Hewett PJ, Flood MP, Pereira S, Whitney GA, Michael M, Tie J, Mukherjee S, Grandori C, Heriot AG, Worthley DL, Ramsay RG, Woods SL. Medium-throughput drug screening of patient-derived organoids from colorectal peritoneal metastases to direct personalized therapy. Clin Cancer Res. 2020;26:3662–70. https://doi.org/10.1158/1078-0432.CCR-20-0073.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Otte J, Dizdar L, Behrens B, Goering W, Knoefel WT, Wruck W, Stoecklein NH, Adjaye J. FGF signalling in the self-renewal of colon cancer organoids. Sci Rep. 2019;9:17365. https://doi.org/10.1038/s41598-019-53907-7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Roerink SF, Sasaki N, Lee-Six H, Young MD, Alexandrov LB, Behjati S, Mitchell TJ, Grossmann S, Lightfoot H, Egan DA, Pronk A, Smakman N, van Gorp J, Anderson E, Gamble SJ, Alder C, van de Wetering M, Campbell PJ, Stratton MR, Clevers H. Intra-tumour diversification in colorectal cancer at the single-cell level. Nature. 2018;556:457–62. https://doi.org/10.1038/s41586-018-0024-3.

    Article  CAS  PubMed  Google Scholar 

  60. Verissimo CS, Overmeer RM, Ponsioen B, Drost J, Mertens S, Verlaan-Klink I, van Gerwen B, van der Ven M, van de Wetering M, Egan DA, Bernards R, Clevers H, Bos JL, Snippert HJ. Targeting mutant RAS in patient-derived colorectal cancer organoids by combinatorial drug screening. eLife. 2016;5:e18489. https://doi.org/10.7554/eLife.18489.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Vlachogiannis G, Hedayat S, Vatsiou A, Jamin Y, Fernández-Mateos J, Khan K, Lampis A, Eason K, Huntingford I, Burke R, Rata M, Koh D-M, Tunariu N, Collins D, Hulkki-Wilson S, Ragulan C, Spiteri I, Moorcraft SY, Chau I, Rao S, Watkins D, Fotiadis N, Bali M, Darvish-Damavandi M, Lote H, Eltahir Z, Smyth EC, Begum R, Clarke PA, Hahne JC, Dowsett M, de Bono J, Workman P, Sadanandam A, Fassan M, Sansom OJ, Eccles S, Starling N, Braconi C, Sottoriva A, Robinson SP, Cunningham D, Valeri N. Patient-derived organoids model treatment response of metastatic gastrointestinal cancers. Science. 2018;359:920–6. https://doi.org/10.1126/science.aao2774.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Yan HHN, Siu HC, Law S, Ho SL, Yue SSK, Tsui WY, Chan D, Chan AS, Ma S, Lam KO, Bartfeld S, Man AHY, Lee BCH, Chan ASY, Wong JWH, Cheng PSW, Chan AKW, Zhang J, Shi J, Fan X, Kwong DLW, Mak TW, Yuen ST, Clevers H, Leung SY. A comprehensive human gastric cancer organoid biobank captures tumor subtype heterogeneity and enables therapeutic screening. Cell Stem Cell. 2018;23:882–897.e11. https://doi.org/10.1016/j.stem.2018.09.016.

    Article  CAS  PubMed  Google Scholar 

  63. Schutgens F, Rookmaaker MB, Margaritis T, Rios A, Ammerlaan C, Jansen J, Gijzen L, Vormann M, Vonk A, Viveen M, Yengej FY, Derakhshan S, de Winter-de Groot KM, Artegiani B, van Boxtel R, Cuppen E, Hendrickx APA, van den Heuvel-Eibrink MM, Heitzer E, Lanz H, Beekman J, Murk J-L, Masereeuw R, Holstege F, Drost J, Verhaar MC, Clevers H. Tubuloids derived from human adult kidney and urine for personalized disease modeling. Nat Biotechnol. 2019;37:303–13. https://doi.org/10.1038/s41587-019-0048-8.

    Article  CAS  PubMed  Google Scholar 

  64. Kim M, Mun H, Sung CO, Cho EJ, Jeon H-J, Chun S-M, Jung DJ, Shin TH, Jeong GS, Kim DK, Choi EK, Jeong S-Y, Taylor AM, Jain S, Meyerson M, Jang SJ. Patient-derived lung cancer organoids as in vitro cancer models for therapeutic screening. Nat Commun. 2019;10:3991. https://doi.org/10.1038/s41467-019-11867-6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Li Z, Qian Y, Li W, Liu L, Yu L, Liu X, Wu G, Wang Y, Luo W, Fang F, Liu Y, Song F, Cai Z, Chen W, Huang W. Human lung adenocarcinoma-derived organoid models for drug screening. iScience. 2020;23:101411. https://doi.org/10.1016/j.isci.2020.101411.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Hill SJ, Decker B, Roberts EA, Horowitz NS, Muto MG, Worley MJ, Feltmate CM, Nucci MR, Swisher EM, Nguyen H, Yang C, Morizane R, Kochupurakkal BS, Do KT, Konstantinopoulos PA, Liu JF, Bonventre JV, Matulonis UA, Shapiro GI, Berkowitz RS, Crum CP, D’Andrea AD. Prediction of DNA repair inhibitor response in short-term patient-derived ovarian cancer organoids. Cancer Discov. 2018;8:1404–21. https://doi.org/10.1158/2159-8290.CD-18-0474.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Kopper O, de Witte CJ, Lõhmussaar K, Valle-Inclan JE, Hami N, Kester L, Balgobind AV, Korving J, Proost N, Begthel H, van Wijk LM, Revilla SA, Theeuwsen R, van de Ven M, van Roosmalen MJ, Ponsioen B, Ho VWH, Neel BG, Bosse T, Gaarenstroom KN, Vrieling H, Vreeswijk MPG, van Diest PJ, Witteveen PO, Jonges T, Bos JL, van Oudenaarden A, Zweemer RP, Snippert HJG, Kloosterman WP, Clevers H. An organoid platform for ovarian cancer captures intra- and interpatient heterogeneity. Nat Med. 2019;25:838–49. https://doi.org/10.1038/s41591-019-0422-6.

    Article  CAS  PubMed  Google Scholar 

  68. Nelson SR, Zhang C, Roche S, O’Neill F, Swan N, Luo Y, Larkin A, Crown J, Walsh N. Modelling of pancreatic cancer biology: transcriptomic signature for 3D PDX-derived organoids and primary cell line organoid development. Sci Rep. 2020;10:2778. https://doi.org/10.1038/s41598-020-59368-7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Tiriac H, Belleau P, Engle DD, Plenker D, Deschênes A, Somerville TDD, Froeling FEM, Burkhart RA, Denroche RE, Jang G-H, Miyabayashi K, Young CM, Patel H, Ma M, LaComb JF, Palmaira RLD, Javed AA, Huynh JC, Johnson M, Arora K, Robine N, Shah M, Sanghvi R, Goetz AB, Lowder CY, Martello L, Driehuis E, LeComte N, Askan G, Iacobuzio-Donahue CA, Clevers H, Wood LD, Hruban RH, Thompson E, Aguirre AJ, Wolpin BM, Sasson A, Kim J, Wu M, Bucobo JC, Allen P, Sejpal DV, Nealon W, Sullivan JD, Winter JM, Gimotty PA, Grem JL, DiMaio DJ, Buscaglia JM, Grandgenett PM, Brody JR, Hollingsworth MA, O’Kane GM, Notta F, Kim E, Crawford JM, Devoe C, Ocean A, Wolfgang CL, Yu KH, Li E, Vakoc CR, Hubert B, Fischer SE, Wilson JM, Moffitt R, Knox J, Krasnitz A, Gallinger S, Tuveson DA. Organoid profiling identifies common responders to chemotherapy in pancreatic cancer. Cancer Res. 2018;19:1112–29. https://doi.org/10.1158/2159-8290.CD-18-0349.

  70. Drost J, Karthaus WR, Gao D, Driehuis E, Sawyers CL, Chen Y, Clevers H. Organoid culture systems for prostate epithelial tissue and prostate cancer tissue. Nat Protoc. 2016;11:347–58. https://doi.org/10.1038/nprot.2016.006.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Puca L, Bareja R, Prandi D, Shaw R, Benelli M, Karthaus WR, Hess J, Sigouros M, Donoghue A, Kossai M, Gao D, Cyrta J, Sailer V, Vosoughi A, Pauli C, Churakova Y, Cheung C, Deonarine LD, McNary TJ, Rosati R, Tagawa ST, Nanus DM, Mosquera JM, Sawyers CL, Chen Y, Inghirami G, Rao RA, Grandori C, Elemento O, Sboner A, Demichelis F, Rubin MA, Beltran H. Patient derived organoids to model rare prostate cancer phenotypes. Nat Commun. 2018;9:2404. https://doi.org/10.1038/s41467-018-04495-z.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Ootani A, Li X, Sangiorgi E, Ho QT, Ueno H, Toda S, Sugihara H, Fujimoto K, Weissman IL, Capecchi MR, Kuo CJ. Sustained in vitro intestinal epithelial culture within a Wnt-dependent stem cell niche. Nat Med. 2009;15:701–6. https://doi.org/10.1038/nm.1951.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Li X, Nadauld L, Ootani A, Corney DC, Pai RK, Gevaert O, Cantrell MA, Rack PG, Neal JT, Chan CW-M, Yeung T, Gong X, Yuan J, Wilhelmy J, Robine S, Attardi LD, Plevritis SK, Hung KE, Chen C-Z, Ji HP, Kuo CJ. Oncogenic transformation of diverse gastrointestinal tissues in primary organoid culture. Nat Med. 2014;20:769–77. https://doi.org/10.1038/nm.3585.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Neal JT, Li X, Zhu J, Giangarra V, Grzeskowiak CL, Ju J, Liu IH, Chiou S-H, Salahudeen AA, Smith AR, Deutsch BC, Liao L, Zemek AJ, Zhao F, Karlsson K, Schultz LM, Metzner TJ, Nadauld LD, Tseng Y-Y, Alkhairy S, Oh C, Keskula P, Mendoza-Villanueva D, De La Vega FM, Kunz PL, Liao JC, Leppert JT, Sunwoo JB, Sabatti C, Boehm JS, Hahn WC, Zheng GXY, Davis MM, Kuo CJ. Organoid modeling of the tumor immune microenvironment. Cell. 2018;175:1972–1988.e16. https://doi.org/10.1016/j.cell.2018.11.021.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Movia D, Bazou D, Volkov Y, Prina-Mello A. Multilayered cultures of NSCLC cells grown at the air-liquid interface allow the efficacy testing of inhaled anti-cancer drugs. Sci Rep. 2018;8:12920. https://doi.org/10.1038/s41598-018-31332-6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Movia D, Bazou D, Prina-Mello A. ALI multilayered co-cultures mimic biochemical mechanisms of the cancer cell-fibroblast cross-talk involved in NSCLC MultiDrug Resistance. BMC Cancer. 2019;19:1–21. https://doi.org/10.1186/s12885-019-6038-x.

    Article  CAS  Google Scholar 

  77. Leene W, Duyzings MJ, van Steeg C. Lymphoid stem cell identification in the developing thymus and bursa of Fabricius of the chick. Z Zellforsch Mikrosk Anat Vienna Austria 1948. 1973;136:521–33. https://doi.org/10.1007/BF00307368.

    Article  CAS  Google Scholar 

  78. Nowak-Sliwinska P, Segura T, Iruela-Arispe ML. The chicken chorioallantoic membrane model in biology, medicine and bioengineering. Angiogenesis. 2014;17:779–804. https://doi.org/10.1007/s10456-014-9440-7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Vu BT, Shahin SA, Croissant J, Fatieiev Y, Matsumoto K, Le-Hoang Doan T, Yik T, Simargi S, Conteras A, Ratliff L, Jimenez CM, Raehm L, Khashab N, Durand J-O, Glackin C, Tamanoi F. Chick chorioallantoic membrane assay as an in vivo model to study the effect of nanoparticle-based anticancer drugs in ovarian cancer. Sci Rep. 2018;8:8524. https://doi.org/10.1038/s41598-018-25573-8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Schmitd LB, Liu M, Scanlon CS, Banerjee R, D’Silva NJ. The chick chorioallantoic membrane in vivo model to assess perineural invasion in head and neck cancer. JoVE J Vis Exp. 2019:e59296. https://doi.org/10.3791/59296.

  81. Janser FA, Ney P, Pinto MT, Langer R, Tschan MP. The Chick Chorioallantoic Membrane (CAM) assay as a three-dimensional model to study autophagy in cancer cells. Bio-Protoc. 2019;9:e3290.

    Google Scholar 

  82. Pawlikowska P, Tayoun T, Oulhen M, Faugeroux V, Rouffiac V, Aberlenc A, Pommier AL, Honore A, Marty V, Bawa O, Lacroix L, Scoazec JY, Chauchereau A, Laplace-Builhe C, Farace F. Exploitation of the chick embryo chorioallantoic membrane (CAM) as a platform for anti-metastatic drug testing. Sci Rep. 2020;10:16876. https://doi.org/10.1038/s41598-020-73632-w.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Mangir N, Raza A, Haycock JW, Chapple C, Macneil S. An improved in vivo methodology to visualise tumour induced changes in vasculature using the chick chorionic allantoic membrane assay. In Vivo. 2018;32:461–72. https://doi.org/10.21873/invivo.11262.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Minami N, Maeda Y, Shibao S, Arima Y, Ohka F, Kondo Y, Maruyama K, Kusuhara M, Sasayama T, Kohmura E, Saya H, Sampetrean O. Organotypic brain explant culture as a drug evaluation system for malignant brain tumors. Cancer Med. 2017;6:2635–45. https://doi.org/10.1002/cam4.1174.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Kenerson HL, Sullivan KM, Seo YD, Stadeli KM, Ussakli C, Yan X, Lausted C, Pillarisetty VG, Park JO, Riehle KJ, Yeh M, Tian Q, Yeung RS. Tumor slice culture as a biologic surrogate of human cancer. Ann Transl Med. 2020;8:114. https://doi.org/10.21037/atm.2019.12.88.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Vaira V, Fedele G, Pyne S, Fasoli E, Zadra G, Bailey D, Snyder E, Faversani A, Coggi G, Flavin R, Bosari S, Loda M. Preclinical model of organotypic culture for pharmacodynamic profiling of human tumors. Proc Natl Acad Sci. 2010;107:8352–6. https://doi.org/10.1073/pnas.0907676107.

    Article  PubMed  PubMed Central  Google Scholar 

  87. Merz F, Gaunitz F, Dehghani F, Renner C, Meixensberger J, Gutenberg A, Giese A, Schopow K, Hellwig C, Schäfer M, Bauer M, Stöcker H, Taucher-Scholz G, Durante M, Bechmann I. Organotypic slice cultures of human glioblastoma reveal different susceptibilities to treatments. Neuro-Oncol. 2013;15:670–81. https://doi.org/10.1093/neuonc/not003.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Martin SZ, Wagner DC, Hörner N, Horst D, Lang H, Tagscherer KE, Roth W. Ex vivo tissue slice culture system to measure drug-response rates of hepatic metastatic colorectal cancer. BMC Cancer. 2019;19:1–14. https://doi.org/10.1186/s12885-019-6270-4.

    Article  CAS  Google Scholar 

  89. Jiang X, Seo YD, Chang JH, Coveler A, Nigjeh EN, Pan S, Jalikis F, Yeung RS, Crispe IN, Pillarisetty VG. Long-lived pancreatic ductal adenocarcinoma slice cultures enable precise study of the immune microenvironment. Onco Targets Ther. 2017;6:e1333210. https://doi.org/10.1080/2162402X.2017.1333210.

    Article  Google Scholar 

  90. Misra S, Moro CF, Del Chiaro M, Pouso S, Sebestyén A, Löhr M, Björnstedt M, Verbeke CS. Ex vivo organotypic culture system of precision-cut slices of human pancreatic ductal adenocarcinoma. Sci Rep. 2019;9:2133. https://doi.org/10.1038/s41598-019-38603-w.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Sivakumar R, Chan M, Shin JS, Nishida-Aoki N, Kenerson HL, Elemento O, Beltran H, Yeung R, Gujral TS. Organotypic tumor slice cultures provide a versatile platform for immuno-oncology and drug discovery. OncoImmunology. 2019;8:e1670019. https://doi.org/10.1080/2162402X.2019.1670019.

    Article  PubMed  PubMed Central  Google Scholar 

  92. Sönnichsen R, Hennig L, Blaschke V, Winter K, Körfer J, Hähnel S, Monecke A, Wittekind C, Jansen-Winkeln B, Thieme R, Gockel I, Grosser K, Weimann A, Kubick C, Wiechmann V, Aigner A, Bechmann I, Lordick F, Kallendrusch S. Individual susceptibility analysis using patient-derived slice cultures of colorectal carcinoma. Clin Colorectal Cancer. 2018;17:e189–99. https://doi.org/10.1016/j.clcc.2017.11.002.

    Article  PubMed  Google Scholar 

  93. Naipal KAT, Verkaik NS, Sánchez H, van Deurzen CHM, den Bakker MA, Hoeijmakers JHJ, Kanaar R, Vreeswijk MPG, Jager A, van Gent DC. Tumor slice culture system to assess drug response of primary breast cancer. BMC Cancer. 2016;16:78. https://doi.org/10.1186/s12885-016-2119-2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Bhatia SN, Ingber DE. Microfluidic organs-on-chips. Nat Biotechnol. 2014;32:760–72. https://doi.org/10.1038/nbt.2989.

    Article  CAS  PubMed  Google Scholar 

  95. Bhise NS, Manoharan V, Massa S, Tamayol A, Ghaderi M, Miscuglio M, Lang Q, Shrike Zhang Y, Shin SR, Calzone G, Annabi N, Shupe TD, Bishop CE, Atala A, Dokmeci MR, Khademhosseini A. A liver-on-a-chip platform with bioprinted hepatic spheroids. Biofabrication. 2016;8:014101. https://doi.org/10.1088/1758-5090/8/1/014101.

    Article  CAS  PubMed  Google Scholar 

  96. Lanz HL, Saleh A, Kramer B, Cairns J, Ng CP, Yu J, Trietsch SJ, Hankemeier T, Joore J, Vulto P, Weinshilboum R, Wang L. Therapy response testing of breast cancer in a 3D high-throughput perfused microfluidic platform. BMC Cancer. 2017;17:1–11. https://doi.org/10.1186/s12885-017-3709-3.

    Article  CAS  Google Scholar 

  97. Jin D, Ma X, Luo Y, Fang S, Xie Z, Li X, Qi D, Zhang F, Kong J, Li J, Lin B, Liu T. Application of a microfluidic-based perivascular tumor model for testing drug sensitivity in head and neck cancers and toxicity in endothelium. RSC Adv. 2016;6:29598–607. https://doi.org/10.1039/C6RA01456A.

    Article  CAS  Google Scholar 

  98. Lu S, Cuzzucoli F, Jiang J, Liang L-G, Wang Y, Kong M, Zhao X, Cui W, Li J, Wang S. Development of a biomimetic liver tumor-on-a-chip model based on decellularized liver matrix for toxicity testing. Lab Chip. 2018;18:3379–92. https://doi.org/10.1039/C8LC00852C.

    Article  CAS  PubMed  Google Scholar 

  99. Eduati F, Utharala R, Madhavan D, Neumann UP, Longerich T, Cramer T, Saez-Rodriguez J, Merten CA. A microfluidics platform for combinatorial drug screening on cancer biopsies. Nat Commun. 2018;9:2434. https://doi.org/10.1038/s41467-018-04919-w.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Holton AB, Sinatra FL, Kreahling J, Conway AJ, Landis DA, Altiok S. Microfluidic biopsy trapping device for the real-time monitoring of tumor microenvironment. PLoS One. 2017;12:e0169797. https://doi.org/10.1371/journal.pone.0169797.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Jenkins RW, Aref AR, Lizotte PH, Ivanova E, Stinson S, Zhou CW, Bowden M, Deng J, Liu H, Miao D, He MX, Walker W, Zhang G, Tian T, Cheng C, Wei Z, Palakurthi S, Bittinger M, Vitzthum H, Kim JW, Merlino A, Quinn M, Venkataramani C, Kaplan JA, Portell A, Gokhale PC, Phillips B, Smart A, Rotem A, Jones RE, Keogh L, Anguiano M, Stapleton L, Jia Z, Barzily-Rokni M, Cañadas I, Thai TC, Hammond MR, Vlahos R, Wang ES, Zhang H, Li S, Hanna GJ, Huang W, Hoang MP, Piris A, Eliane J-P, Stemmer-Rachamimov AO, Cameron L, Su M-J, Shah P, Izar B, Thakuria M, LeBoeuf NR, Rabinowits G, Gunda V, Parangi S, Cleary JM, Miller BC, Kitajima S, Thummalapalli R, Miao B, Barbie TU, Sivathanu V, Wong J, Richards WG, Bueno R, Yoon CH, Miret J, Herlyn M, Garraway LA, Allen EMV, Freeman GJ, Kirschmeier PT, Lorch JH, Ott PA, Hodi FS, Flaherty KT, Kamm RD, Boland GM, Wong K-K, Dornan D, Paweletz CP, Barbie DA. Ex vivo profiling of PD-1 blockade using organotypic tumor spheroids. Cancer Discov. 2018;8:196–215. https://doi.org/10.1158/2159-8290.CD-17-0833.

    Article  CAS  PubMed  Google Scholar 

  102. Deng J, Wang ES, Jenkins RW, Li S, Dries R, Yates K, Chhabra S, Huang W, Liu H, Aref AR, Ivanova E, Paweletz CP, Bowden M, Zhou CW, Herter-Sprie GS, Sorrentino JA, Bisi JE, Lizotte PH, Merlino AA, Quinn MM, Bufe LE, Yang A, Zhang Y, Zhang H, Gao P, Chen T, Cavanaugh ME, Rode AJ, Haines E, Roberts PJ, Strum JC, Richards WG, Lorch JH, Parangi S, Gunda V, Boland GM, Bueno R, Palakurthi S, Freeman GJ, Ritz J, Nicholas Haining W, Sharpless NE, Arthanari H, Shapiro GI, Barbie DA, Gray NS, Wong K-K. CDK4/6 inhibition augments antitumor immunity by enhancing T-cell activation. Cancer Discov. 2018;8:216–33. https://doi.org/10.1158/2159-8290.CD-17-0915.

    Article  CAS  PubMed  Google Scholar 

  103. Lee SWL, Adriani G, Ceccarello E, Pavesi A, Tan AT, Bertoletti A, Kamm RD, Wong SC. Characterizing the role of monocytes in T cell cancer immunotherapy using a 3D microfluidic model. Front Immunol. 2018;9:416. https://doi.org/10.3389/fimmu.2018.00416.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Pavesi A, Tan AT, Koh S, Chia A, Colombo M, Antonecchia E, Miccolis C, Ceccarello E, Adriani G, Raimondi MT, Kamm RD, Bertoletti A. A 3D microfluidic model for preclinical evaluation of TCR-engineered T cells against solid tumors. JCI Insight. 2017;2:e89762. https://doi.org/10.1172/jci.insight.89762.

    Article  PubMed Central  Google Scholar 

  105. Paek J, Park SE, Lu Q, Park K-T, Cho M, Oh JM, Kwon KW, Yi Y, Song JW, Edelstein HI, Ishibashi J, Yang W, Myerson JW, Kiseleva RY, Aprelev P, Hood ED, Stambolian D, Seale P, Muzykantov VR, Huh D. Microphysiological engineering of self-assembled and perfusable microvascular beds for the production of vascularized three-dimensional human microtissues. ACS Nano. 2019;13:7627–43. https://doi.org/10.1021/acsnano.9b00686.

    Article  CAS  PubMed  Google Scholar 

  106. Jarvis M, Arnold M, Ott J, Pant K, Prabhakarpandian B, Mitragotri S. Microfluidic co-culture devices to assess penetration of nanoparticles into cancer cell mass. Bioeng Transl Med. 2017;2:268–77. https://doi.org/10.1002/btm2.10079.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Sobrino A, Phan DTT, Datta R, Wang X, Hachey SJ, Romero-López M, Gratton E, Lee AP, George SC, Hughes CCW. 3D microtumors in vitro supported by perfused vascular networks. Sci Rep. 2016;6:31589. https://doi.org/10.1038/srep31589.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Seo BR, DelNero P, Fischbach C. In vitro models of tumor vessels and matrix: engineering approaches to investigate transport limitations and drug delivery in cancer. Adv Drug Deliv Rev. 2014;69–70:205–16. https://doi.org/10.1016/j.addr.2013.11.011.

    Article  CAS  PubMed  Google Scholar 

  109. Choi Y, Hyun E, Seo J, Blundell C, Kim HC, Lee E, Lee SH, Moon A, Moon WK, Huh D. A microengineered pathophysiological model of early-stage breast cancer. Lab Chip. 2015;15:3350–7. https://doi.org/10.1039/C5LC00514K.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Hassell BA, Goyal G, Lee E, Sontheimer-Phelps A, Levy O, Chen CS, Ingber DE. Human organ chip models recapitulate orthotopic lung cancer growth, therapeutic responses, and tumor dormancy in vitro. Cell Rep. 2017;21:508–16. https://doi.org/10.1016/j.celrep.2017.09.043.

    Article  CAS  PubMed  Google Scholar 

  111. Hidalgo M, Amant F, Biankin AV, Budinská E, Byrne AT, Caldas C, Clarke RB, de Jong S, Jonkers J, Mælandsmo GM, Roman-Roman S, Seoane J, Trusolino L, Villanueva A. Patient-derived xenograft models: an emerging platform for translational cancer research. Cancer Discov. 2014;4:998–1013. https://doi.org/10.1158/2159-8290.CD-14-0001.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Invrea F, Rovito R, Torchiaro E, Petti C, Isella C, Medico E. Patient-derived xenografts (PDXs) as model systems for human cancer. Curr Opin Biotechnol. 2020;63:151–6. https://doi.org/10.1016/j.copbio.2020.01.003.

    Article  CAS  PubMed  Google Scholar 

  113. Linxweiler J, Hajili T, Körbel C, Berchem C, Zeuschner P, Müller A, Stöckle M, Menger MD, Junker K, Saar M. Cancer-associated fibroblasts stimulate primary tumor growth and metastatic spread in an orthotopic prostate cancer xenograft model. Sci Rep. 2020;10:12575. https://doi.org/10.1038/s41598-020-69424-x.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Ben-David U, Ha G, Tseng Y-Y, Greenwald NF, Oh C, Shih J, McFarland JM, Wong B, Boehm JS, Beroukhim R, Golub TR. Patient-derived xenografts undergo mouse-specific tumor evolution. Nat Genet. 2017;49:1567–75. https://doi.org/10.1038/ng.3967.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Erstad DJ, Sojoodi M, Taylor MS, Ghoshal S, Razavi AA, Graham-O’Regan KA, Bardeesy N, Ferrone CR, Lanuti M, Caravan P, Tanabe KK, Fuchs BC. Orthotopic and heterotopic murine models of pancreatic cancer and their different responses to FOLFIRINOX chemotherapy. Dis Model Mech. 2018;11:dmm034793. https://doi.org/10.1242/dmm.034793.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Lai Y, Wei X, Lin S, Qin L, Cheng L, Li P. Current status and perspectives of patient-derived xenograft models in cancer research. J Hematol Oncol. 2017;10:106. https://doi.org/10.1186/s13045-017-0470-7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Tian H, Lyu Y, Yang Y-G, Hu Z. Humanized rodent models for cancer research. Front Oncol. 2020;10:1696. https://doi.org/10.3389/fonc.2020.01696.

    Article  PubMed  PubMed Central  Google Scholar 

  118. Jespersen H, Lindberg MF, Donia M, Söderberg EMV, Andersen R, Keller U, Ny L, Svane IM, Nilsson LM, Nilsson JA. Clinical responses to adoptive T-cell transfer can be modeled in an autologous immune-humanized mouse model. Nat Commun. 2017;8:707. https://doi.org/10.1038/s41467-017-00786-z.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Clohessy JG, Pandolfi PP. Mouse hospital and co-clinical trial project—from bench to bedside. Nat Rev Clin Oncol. 2015;12:491–8. https://doi.org/10.1038/nrclinonc.2015.62.

    Article  PubMed  Google Scholar 

  120. Clohessy JG, Pandolfi PP. The mouse hospital and its integration in ultra-precision approaches to cancer care. Front Oncol. 2018;8:340. https://doi.org/10.3389/fonc.2018.00340.

    Article  PubMed  PubMed Central  Google Scholar 

  121. Koga Y, Ochiai A. Systematic review of patient-derived xenograft models for preclinical studies of anti-cancer drugs in solid tumors. Cells. 2019;8:418. https://doi.org/10.3390/cells8050418.

    Article  CAS  PubMed Central  Google Scholar 

  122. Bangi E, Murgia C, Teague AGS, Sansom OJ, Cagan RL. Functional exploration of colorectal cancer genomes using Drosophila. Nat Commun. 2016;7:13615. https://doi.org/10.1038/ncomms13615.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Das TK, Esernio J, Cagan RL. Restraining network response to targeted cancer therapies improves efficacy and reduces cellular resistance. Cancer Res. 2018;78:4344–59. https://doi.org/10.1158/0008-5472.CAN-17-2001.

    Article  CAS  PubMed  Google Scholar 

  124. Bangi E, Ang C, Smibert P, Uzilov AV, Teague AG, Antipin Y, Chen R, Hecht C, Gruszczynski N, Yon WJ, Malyshev D, Laspina D, Selkridge I, Rainey H, Moe AS, Lau CY, Taik P, Wilck E, Bhardwaj A, Sung M, Kim S, Yum K, Sebra R, Donovan M, Misiukiewicz K, Schadt EE, Posner MR, Cagan RL. A personalized platform identifies trametinib plus zoledronate for a patient with KRAS-mutant metastatic colorectal cancer. Sci Adv. 2019;5:eaav6528. https://doi.org/10.1126/sciadv.aav6528.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Benjamin D. Hopkins .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2022 Springer Nature Switzerland AG

About this chapter

Check for updates. Verify currency and authenticity via CrossMark

Cite this chapter

Claridge, S.E., Cavallo, JA., Hopkins, B.D. (2022). Patient-Derived In Vitro and In Vivo Models of Cancer. In: Laganà, A. (eds) Computational Methods for Precision Oncology. Advances in Experimental Medicine and Biology, vol 1361. Springer, Cham. https://doi.org/10.1007/978-3-030-91836-1_12

Download citation

Publish with us

Policies and ethics